Open Access

Current landscape and future directions of therapeutic approaches for adenoid cystic carcinoma of the salivary glands (Review)

  • Authors:
    • Katarzyna Stawarz
    • Monika Durzynska
    • Adam Gałązka
    • Anna Gorzelnik
    • Jakub Zwolinski
    • Monika Paszkowska
    • Karolina Bieńkowska‑Pluta
    • Magdalena Misiak‑Galazka
  • View Affiliations

  • Published online on: January 22, 2025     https://doi.org/10.3892/ol.2025.14899
  • Article Number: 153
  • Copyright: © Stawarz et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adenoid cystic carcinoma (ACC) of the salivary glands is the second most common type of salivary gland cancer, and is characterized by a poor prognosis and an unclear pathology. The incidence of ACC is rare, as it accounts for 10‑15% of all salivary gland tumors and affects mainly patients aged between 50 and 60 years. The annual incidence rate is estimated to be ~4.5 cases per 100,000 individuals. Due to its rarity and the use of contaminated cell lines in previous investigations, the precise etiological factors underlying ACC remain poorly understood. Current treatment modalities, typically involving surgery with or without postoperative radiotherapy, often prove unsatisfactory due to the potential for local recurrence and delayed distant metastases, which may manifest 3‑5 years after treatment and constitute the primary failure of existing therapeutic approaches. The indolent growth pattern, along with perineural and perivascular invasion, is potentially responsible for the delayed onset of metastases. No effective systemic therapy has been established so far. Therefore, the management of ACC represents a significant therapeutic challenge. Exploring the molecular characteristics of ACC, including the reasons behind its propensity for perineural invasion and its potential correlation with the immune system, offers promising strategies for managing ACC and could open up novel pathways for future therapeutic interventions. Currently, the use of immunotherapy in ACC treatment has shown limited effectiveness. While the exact mechanism underlying the lack of response to immunotherapy in ACC remains unknown, the low levels of tumor‑infiltrating lymphocytes in these tumors may contribute to this resistance. Therefore, identifying novel targets to enhance the immune response against tumor cells is essential. The present review provides an update on clinical studies and explores novel therapeutic targets that could be effective in the therapeutic management of ACC.

Introduction

Adenoid cystic carcinoma (ACC) is a rare malignancy that arises from cells within the salivary glands, although its precise cellular origin remains unclear (1). ACC comprises ~1% of all types of head and neck cancer, positioning itself as the second most common salivary gland cancer after mucoepidermoid carcinoma (2). ACC predominantly arises in the minor salivary glands, a location which accounts for >50% of all ACC cases, and is primarily localized within the oral cavity, hard palate, throat mucosa or paranasal sinuses (3). While the salivary glands are the most common site for ACC, this type of cancer can also arise in other locations, such as the breast, lungs or the Bartholin glands. ACC is particularly rare in the breast, accounting for <1% of all breast cancer cases (4). Unlike the salivary gland subtype, breast ACC, while slow growing, generally has a favorable prognosis (4). Pulmonary ACC, by contrast, shares few similarities with its salivary counterpart. Although the histological patterns of these tumors are similar, perineural invasion is notably less common in pulmonary ACC compared to salivary ACC. Additionally, pulmonary ACC has a slightly better prognosis (5). Surgical resection remains the treatment of choice for pulmonary ACC, as no effective systemic therapy is currently established (5). Due to its non-specific clinical presentation, ACC in the Bartholin glands often goes unnoticed and is therefore frequently diagnosed at an advanced stage (6). ACC exhibits a propensity for perineural invasion and distant metastases, which may manifest years after initial treatment (7). Surgery followed by radiotherapy remains the cornerstone of ACC treatment (8). Due to the use of contaminated cell lines in numerous previous research studies (9,10) further investigation is needed to elucidate the pathogenesis of ACC and identify potential therapeutic targets (11). The ACC2, ACC3 and ACCM cell lines were primarily contaminated with cervical cancer cells, while the ACCS cell line was composed of T24 urinary bladder cancer cells (11).

Several recently published papers (1214) have provided new insights into potential therapeutic targets for managing ACC. In the present review, a comprehensive literature search of publications from January 2019 to April 2024 in the PubMed (http://www.ncbi.nlm.nih.gov/pubmed) and Cochrane Library (https://www.cochranelibrary.com) databases was conducted. Case reports were excluded from the analysis. The primary aim of the present review was to elucidate advancements in the treatment of ACC, while also highlighting potential pathways for therapeutic interventions in patients with ACC. Articles focusing on therapeutic modalities were selected, with particular emphasis on novel possible therapies validated by randomized controlled trials.

Epidemiology

Current data suggests that ACC occurs more frequently in females, with a distribution of 60% in females compared with 40% in males (15). The overall incidence of ACC accounts for ~4.5 cases per 100,000 individuals (16), representing ~1% of all types of head and neck cancer and ~10% of all salivary gland tumors (17). ACC, originating from the mucous glands, can occur in various body sites beyond the salivary glands (18). A large proportion of the available literature indicates that minor salivary glands represent the most common site for ACC, accounting for >50% of cases. The palate is the most frequently affected location, although ACC can also develop in the tongue, paranasal sinuses, nasopharynx, larynx and lacrimal glands (19,20). Among the major salivary glands, the parotid gland is the most prevalent site for ACC (21). Additionally, ACC may arise in the breast, tracheobronchial tree, lungs, prostate, esophagus, skin, Bartholin glands and cervix (22). Although this malignancy can occur at any age, it is most commonly diagnosed in patients between 50 and 60 years of age (23).

Etiology

Due to the rarity of ACC, the precise etiology and pathogenesis remain poorly understood (24). While smoking and alcohol intake are known risk factors for other types of cancer in the head and neck area, especially squamous cell carcinoma, they have not been confirmed as risk factors for ACC (25). Several genetic and chromosomal alterations, including TP53, NOTCH1, NOTCH2 or loss of the CDKN2A/CDKN2B locus, may be involved in ACC pathogenesis, with a recurrent t(6;9)(q23;p23) translocation resulting in a fusion between the MYB proto-oncogene, transcription factor (MYB) and nuclear factor I/B (NFIB) genes, considered to be a genetic hallmark of ACC (26). Nonetheless, the exact causes of ACC, including potential risk factors, require additional research.

Clinical presentation and diagnosis

The symptoms of ACC can vary depending on the location of the tumor (27), with a lack of symptoms reported if the tumor is located in the paranasal sinuses or dysphagia if it is located on the hard palate, the base of the tongue or the throat mucosa (28). Dyspnea, coughing, hoarseness or wheezing may occur if the minor salivary glands of the upper aerodigestive tract are involved (29). In advanced stages of the disease, dull pain and altered sensation of the tongue, palate or face may occur, which are manifestations of perineural invasion (PNI) of the local nerves, a characteristic phenomenon of ACC (30). PNI is relatively common, affecting ~43.2% of cases and is considered an independent factor for a poor prognosis (31). Despite its slow development, ACC is considered an aggressive tumor that can readily invade the surrounding tissues. Other factors that can influence the risk of distant metastases include a solid histology, a tumor size >3 cm and the involvement of loco-regional lymph nodes (32). Distant metastases are not uncommon and tend to spread via the perivascular route, typically affecting the lungs, followed by the bones and liver (33). As a result, local recurrences (60%) and distant metastases (40%) are frequent, and can occur even decades after definitive treatment (34). The 5-year overall survival rate ranges from 55 to 70%, which is higher compared with that of other sinonasal malignancies, but the overall survival rate drops to 40% at 10 years and further to 15% at 20 years after diagnosis. Therefore, extended follow-up for at least 15 years, if not lifelong follow-up, is necessary (35). Distinguishing salivary ACC from other malignant salivary tumors requires advanced pathological skills. ACC shares histopathological features with other salivary gland tumors, such as polymorphous adenocarcinoma and basal cell adenocarcinoma (27). The cribriform, tubular and solid growth patterns observed in ACC can occasionally resemble other malignancies, including pleomorphic adenoma or polymorphous low-grade adenocarcinoma, leading to diagnostic confusion (36). While the classic cribriform pattern is often recognizable, the tubular and solid variants can be more difficult to distinguish from other salivary gland tumors, particularly when these patterns dominate the tumor (36). Therefore, the role of expert pathological review in the diagnosis of salivary ACC is crucial due to the propensity of the tumor for late local recurrence and distant metastasis. In such cases, experienced pathologists could accurately evaluate biopsy samples to confirm the presence of recurrent or metastatic ACC and distinguish it from other possible malignancies or benign conditions. Expert pathological review would serve as a quality assurance measure, and provide a second opinion to confirm initial diagnoses and prevent diagnostic errors. The role of molecular diagnostics in salivary ACC diagnosis is also critical for distinguishing the tumor from other salivary gland tumors. Specifically, the MYB-NFIB gene fusion, a molecular hallmark of ACC, aids in differentiating it from other tumors, such as Warthin's tumor (37). Additionally, while immunohistochemical staining can assist in the diagnosis, markers such as S-100, CK-7, CK-17 or SOX10 are not entirely specific to ACC and may overlap with those of other salivary gland tumors. Therefore, careful interpretation of staining patterns in conjunction with histological findings, is essential. The definitive diagnosis of salivary ACC remains challenging due to the histological diversity of types of salivary gland cancer. This complexity is underscored by the 2022 World Health Organization classification of salivary gland tumors, which introduces new malignant entities such as microsecretory carcinoma and sclerosing microcystic adenocarcinoma (38).

Clinical management

Current treatment options for ACC typically involve surgical resection followed by postoperative radiotherapy, which appears to be a feasible approach for achieving locoregional control in early stage disease (39). In a study by Ishida et al (40), which involved 58 cases of ACC treated solely with surgical excision, the 10-, 20- and 25-year survival rates were 63.7, 27.3 and 20%, respectively. Although surgery remains the preferred therapeutic option for patients with ACC, it often presents significant challenges due to the tumor's location, particularly when it arises from the minor salivary glands in areas such as the paranasal sinuses. Radical resection can be difficult to achieve, which underscores the need for postoperative radiotherapy to compensate for incomplete tumor removal (41). It was previously reported that patients with ACC who did not receive postoperative radiotherapy were >13 times more likely to experience local recurrence (42). Nevertheless, radiotherapy alone is seldom used and is usually dedicated for patients with advanced or recurrent disease (Fig. 1) (43). The frequent incidence of local recurrence and distant metastasis, even years after completion of treatment, highlights the ineffectiveness of these therapies. Furthermore, neither the National Comprehensive Cancer Network (NCCN) (8) nor the American Society of Clinical Oncology (44) provides specific guidelines for effective chemotherapeutic regimens in the management of ACC. In palliative cases, chemotherapeutic regimens typically include cisplatin and 5-fluorouracil, or combination therapies such as cisplatin, doxorubicin and cyclophosphamide (CAP) (45). However, in the use of monotherapy, agents such as cisplatin, mitoxantrone, epirubicin, vinorelbine, paclitaxel and gemcitabine may be employed (46). Nevertheless, the effectiveness of these chemotherapeutics remains limited, since these drugs are reported to have no or only slight effects on the prognosis of the patient (46). According to data from the available literature, the response rate for CAP is estimated to be between 18 and 31% (47). In a phase II study involving cisplatin and docetaxel, which predominantly included patients with ACC, as well as other types of salivary gland cancer, the median duration of response was 6.8 months. The median progression-free survival time was 9.4 months and the overall survival time was ~28.2 months (48). Nevertheless, the data confirmed that chemotherapy has limited effectiveness in treating ACC and is primarily used as a palliative approach.

Figure 1.

Schematic representation of (A) the screening process and (B) therapeutic strategies for addressing recurrence and metastasis in adenoid cystic carcinoma (ACC), as outlined by the National Comprehensive Cancer Network (NCCN) guidelines. ACC, adenoid cystic carcinoma; RT, radiotherapy.

Current and novel therapies

Therapeutic management of ACC

Currently, there is no effective systemic therapy for managing ACC of the salivary glands, particularly in advanced stages or for inoperable tumors. This emphasizes the need to explore new treatment strategies, especially those incorporating targeted therapies, to improve the management of late-stage ACC. Although no new systemic therapies for managing ACC have recently been approved, the primary goal of this review is to outline the current therapeutic targets and describe ongoing clinical trials (Table I) that are exploring potential treatment options for ACC, while also providing data on concluded clinical trials (Table II).

Table I.

List of clinical trials evaluating agents for adenoid cystic carcinoma treatment.

Table I.

List of clinical trials evaluating agents for adenoid cystic carcinoma treatment.

Clinical trial no. Drug name Drug target Phase No. of cases Primary endpoint Estimated study completion date
NCT04974866 EGFR-TKIs EGFR-TKI II 20 ORR by RECIST 2026-07-31
NCT06118086 REM-422 MYB mRNA degrader I 65 ORR by RECIST 2026-06-01
NCT04973683 AL-107 NOTCH inhibition I 14 ORR by RECIST 2024-12-15
NCT05774899 CB-103 with either lenvatinib or abemaciclib CB-103, an oral NOTCH pathway, inhibitor; abemaciclib a CDK4/6 inhibitor; lenvatinib, a VEGFR TKI I/II 34 ORR by RECIST 2026-06-01
NCT06322576 177Lu-PSMA Human PSMA-targeting ligand, conjugated to the β-emitting radioisotope 177Lu II 10 ORR by RECIST 2035-12
NCT02780310 Lenvatinib TKI II 33 ORR by RECIST 2025-05
NCT02098538 Regorafenib TKI II 38 ORR by RECIST 2025-03
NCT06199453 177Lu vipivotide tetraxetan Human PSMA-targeting ligand, conjugated to the β-emitting radioisotope 177Lu II 32 ORR by RECIST 2027-11
NCT05074940 Amivantamab EGFR-MET bispecific antibody II 18 ORR by RECIST 2028-08-05
NCT04209660 Lenvatinib and pembrolizumab VEGFR inhibitor and programmed death receptor 1 inhibitor II 64 ORR by RECIST 2024-12
NCT05930951 OBT076 with or without balstilimab targeting PD1 CD205/Ly75-directed antibody-drug conjugate-targeting the CD205/Ly75 molecule, also known as DEC-205 (Dendritic and Epithelial Cell-205). I 32 ORR by RECIST 2027-09
NCT03146650 Nivolumab and ipilimumab Nivolumab, programmed death-ligand 1; ipilimumab, cytotoxic T cell antigen 4 II 25 ORR by RECIST 2025-08-11
NCT05010629 9-ING-41 with carboplatin 9-ING-41, a GSK-3β inhibitor; carboplatin chemotherapy II 35 ORR by RECIST 2025-08-30
NCT05194072 SGN-B7H4V B7-H4 I 430 ORR by RECIST 2027-01-31
NCT03556228 VMD-928 Tropomyosin receptor A inhibitor I 74 ORR by RECIST 2025-12
NCT04140526 ONC-392 and pembrolizumab Humanized anti-CTLA4 IgG1 monoclonal antibody I/II 914 ORR by RECIST 2024-12-31
NCT04249947 P-PSMA-101 CAR-T cells and rimiducid acting as rapamycin analog P-PSMA-101 CAR-T cells I 60 ORR by RECIST 2036-09

[i] EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; 177Lu, lutetium Lu 177; MYB, MYB proto-oncogene, transcription factor; PSMA, prostate-specific membrane antigen; ORR, objective response rate; RECIST, Response Evaluation Criteria in Solid Tumors.

Table II.

List of clinical trials with published results.

Table II.

List of clinical trials with published results.

Drug No. of cases Biological target Primary endpoint Trial result Clinical trial no. URL
Regorafenib 38 VEGFR1, VEGFR2, VEGFR3, KIT, RET, BRAF and FGFR1 PFS, ≥6 months ORR (CR and PR) for 6 months Not reported NCT02098538 https://clinicaltrials.gov/ct2/show/NCT02098538.
Vorinostat 30 HDAC ORR, up to 6 months - PR, 1 patient (3%) with response   duration of ≥11.2 months. NCT01175980 https://clinicaltrials.gov/ct2/show/
- SD, 25 patients (83%).
- Median PFS, 12.7 months.
- PFS >1 year, 6 patients (20%).
- Median OS, not reached.
Sorafenib 23 Serine/threonine PFS, 12 months - PFS at 12 months, 46.2%. Eudra CT2008- https://ichgcp.net/eu-clinical-trials-
kinases c-Raf/b-Raf, Secondary endpoints: - Median PFS, 11.3 months. 000066-22 registry/trial/2008-000066-22/results
VEGFR2, VEGFR3, ORR, OS and toxicity - Median OS, 19.6 months.
PDGFR-β, FMS-like - PR, 2 of 19 patients (11%).
tyrosine kinase 3, - SD, 13 of 19 patients (68%).
c-kit and p38α - PD, 4 of 19 patients (21%).
Cetuximab 23 EGFR CR, PR and SD - No ORR. https://clinicaltrials.gov/study/
- SD, 20 of 23 patients (87%). NCT00509002
Cetuximab + 49 EGFR Toxicity of the Not reported. NCT01192087 https://clinicaltrials.gov/study/
intensity combined therapy NCT01192087?cond=NCT01192087
modulated composed of RT + &rank=1
radiation cetuximab
therapy Secondary endpoints:
ORR, PFS and OS
Cetuximab + 21 EGFR PFS - For locally advanced ACC EudraCT 2006- https://www.clinicaltrialsregister.eu/
RT + cisplatin Secondary outcomes:   (n=9): Median PFS, 64 months; 001694-23 ctr-search/search?query=2006-
or Cetuximab + ORR, OS   CR, 2 (22%); 2 PR (22%); 5 SD 001694-23
cisplatin and   (55.6%); no PD (0%); and OS
5-FU   was not reached.
- For metastatic ACC (n=12):
  Median PFS, 13 months;
  maximum PFS, 48 months; 5 PR
  (42%); 7 SD (58%); no PD (0%);
  and OS, 24 months.
Dovitinib 20 VEGFR1, VEGFR2, ORR and SD rate Not reported NCT01678105 https://clinicaltrials.gov/study/NCT01
VEGFR3, FGFR1, 678105?cond=NCT01678105&rank=1
FGFR2, FGFR3 and
PDGFR- β
Dovitinib 21 VEGFR1, VEGFR2, ORR - PR, 2 of 19 evaluable patients NCT01524692 https://clinicaltrials.gov/study/
VEGFR3, FGFR1,   (10.5%). NCT01524692?cond=NCT01524692
FGFR2, FGFR3 and - SD >6 months, 9 patients (43%). &rank=1
PDGFR- β - Total SD, 15 (71%).
- 6 patients with shorter follow-up
  did not show progression.
- 4 patients (19%) progressed
  early <4 months.
Axitinib 33 VEGFR1, VRGFR2, ORR - PR, 3 of 33 patients (9%). NCT01558661 https://clinicaltrials.gov/study/
VEGFR3 and c-kit - SD, 25 patients (76%). NCT01558661?cond=NCT01558661
- SD ≥6 months, 11 patients (33%) &rank=1
Bortezomib + 24 26S proteasome, ORR - Bortezomib only: No objective NCT00077428 https://clinicaltrials.gov/study/
doxorubicin NF-κB   response; SD, 15 of 21 evaluable NCT00077428?cond=NCT00077428
  patients (71%); median PFS, &rank=1
  6.4 months; and OS 21 months.
- Doxorubicin added: PR, 1 of
 10 evaluable patients (10%); and
  SD, 6 (60%).
Bortezomib + 10 26S proteasome, ORR and SD rate Not reported NCT00581360 https://clinicaltrials.gov/study/NCT00
doxorubicin NF-κB 581360?cond=NCT00581360&rank=1
Nelfinavir 15 MAPK, PI3K/Akt PFS and ORR - No objective responses. NCT01065844 https://clinicaltrials.gov/study/
signaling pathway - SD, 7 patients (47%). NCT01 065844?cond=NCT01065844&
- SD ≥6 months, 2 (13%). rank=1
- PD, 9 of 12 assessable
  patients (75%).
- Median PFS, 5.5 months.
Sunitinib 14 VEGFR1, VEGFR2, ORR - No objective responses. NCT00886132 https://clinicaltrials.gov/study/
VEGFR3, c-kit, - SD, 11 patients (79%). NCT00886132?cond=NCT00886132&
PDGFR-α, - PD, 2 patients (14%). rank=1
PDGFR- β, RET - Median OS, 18.7 months.
and FLT3 - Median time to progression,
  7.2 months.
MK 2206 19 AKT ORR Not reported NCT01604772 https://clinicaltrials.gov/study/
NCT01604772?cond=NCT01604772&
rank=1
Imatinib 10 c-kit ORR - No objective responses.
- SD, 2 (20%) for ≥6 months.
Dasatinib 40+ c-kit ORR, PFS - No objective responses. NCT00859937 https://clinicaltrials.gov/study/
- PR, 0 patients (0%). NCT00859937
- SD, 21 patients (52%).
- Median PFS, 4.8 months.
Lapatinib 19+ EGFR, ORR - No objective responses. NCT00095563 https://clinicaltrials.gov/study/
erbB2(HER2) - SD, 15 patients (79%). NCT00095563
- SD for ≥6 months,
  9 patients (47%).
Gefitinib 18+ EGFR ORR - No objective responses. https://ichgcp.net/clinical-trials-
- SD ≥9 months, 7 patients (38%). registry/NCT00509002
- Median PFS, 4.3 months.
- Median OS, 25.9 months.
Everolimus 34 mTOR PFS rate at 4 months. - 4-month PFS probability, 65.5% NCT01152840 https://ichgcp.net/clinical-trials-
Secondary endpoint: - No objective responses. registry/NCT01192087
ORR - SD, 27 patients (79%).
- SD ≥6 months, 13 patients (38%).

[i] ACC, adenoid cystic carcinoma; ORR, objective response rate, PFS, progression-free survival; PR, partial response; SD, stable disease; OS, overall survival; PD, progressive disease; CR, complete response; RT, radiotherapy.

Database search strategy and selection criteria

An extensive literature search was conducted using the PubMed and Cochrane Library databases. The search strategy utilized combined Medical Subject Headings terms and key words, including: ‘adenoid cystic carcinoma of a salivary gland’ or ‘ACC’, ‘salivary adenoid cystic carcinoma’, and ‘SACC’. There were no restrictions on language, geographical region, patient age or follow-up duration. Prospective or retrospective clinical and animal studies were included in the present review. The exclusion criteria were as follows: Editorials, letters, reviews, case reports, lack of relevant outcome reporting and duplicate publications.

Vascular endothelial growth factor (VEGF)

The high expression of VEGF in over two-thirds of patients with ACC has been identified as a potentially promising therapeutic target (49). Molecular studies on ACC have also shown that high MYB expression levels are correlated with increased VEGF expression levels (5052). Despite this correlation, preclinical studies using the VEGF inhibitor regorafenib, which showed successful results by inhibiting ACC cell migration and intravascular cancer migration, did not translate into clinical benefit for patients with ACC in human trials (53). Similar unsuccessful outcomes were observed in studies assessing the effectiveness of sunitinib or nintedanib in patients with ACC (54,55). However, lenvatinib and axitinib showed relatively higher objective response rates, with 11–16 and 9–17%, respectively (56). In a phase II clinical trial with axitinib and avelumab in patients with recurrent disease, the partial response was confirmed in 28 out of 40 enrolled patients, with a median overall survival time estimated at 16.6 months. However, the aforementioned study reached its primary end point with 4 partial responses in 28 evaluable patients (57,58). Another study conducted on lenvatinib use among patients with recurrent or metastatic ACC demonstrated a partial response in 5 patients only among the 33 enrolled and 32 evaluable for the primary endpoint. A large proportion of patients (24) had stable disease; however, the severe side effects, including hypertension or oral pain, resulted in treatment discontinuation (59). Locati et al (60) evaluated lenvatinib in patients with recurrent or metastatic ACC and found only three partial responses among 26 participants, with a median overall survival time of 27 months, a median progression-free survival time of 9.1 months and a median duration of response of 3.1 months. In another study assessing the impact of VEGFR2 inhibition in recurrent or metastatic ACC, rivoceranib demonstrated limited effectiveness in 72 evaluated patients, with an overall response rate of 15.3%, a median duration of response of 14.9 months and a median progression-free survival time of 9.0 months (61). A clinical trial evaluating the effectiveness of cabozantinib among patients with salivary gland cancer, including ACC, reported high levels of toxicity and was closed prematurely (59). Additionally, in another study, only 1 out of 15 patients with ACC achieved a partial response, which was comparable to the response rates observed in other types of salivary gland cancer (62). Thus, even though VEGFR is highly expressed in ACC tissue, it does not appear to be an effective therapeutic target.

Epidermal growth factor receptor (EGFR)

EGFR has been found to be overexpressed in >85% of ACC cases (63). However, therapies targeting EGFR inhibition have shown limited effectiveness in managing ACC. The EGFR monoclonal antibodies act by inhibition of phosphorylation and cellular signaling, and support tumor clearance through antibody-dependent cellular cytotoxicity (ADCC) (64). Previous studies on cetuximab, gefitinib and lapatinib inhibiting both EGFR and HER2 did not change the disease course substantially in patients with ACC (15,65). In a study by Bossi et al (66), cetuximab contributed to disease stabilization for >6 months. Despite the increased expression levels of EGFR in ACC, studies investigating EGFR inhibition have shown no objective responses (6769). Nevertheless, in a study conducted by Chew et al (70), the co-administration of prochlorperazine (PCZ) with an EGFR inhibitor was investigated as a way to enhance the availability of surface EGFR for antibody binding, thereby improving ADCC. The inhibition of dynamin by PCZ and clathrin-mediated endocytosis allows for increased EGFR expression on the surface of cancer cells, potentially leading to enhanced ADCC and an improved response in patients with recurrent or metastatic ACC (71).

Fibroblast growth factor receptor 1 (FGFR1)

The increased expression of FGFR1 in ACC contributed to research on the potential FGFR1 inhibitors, which may decrease cancer cell proliferation (72). In an experimental animal model of ACC, use of dovitinib, a multi-kinase inhibitor, resulted in slowed disease progression (73). These findings were subsequently validated in a study on dovitinib in patients with ACC conducted by Dillon et al (74), which observed a partial response in 2 patients and disease stabilization in 65% of included patients (71). Despite achieving disease stabilization, 67% of the patients eventually experienced cancer progression, with an overall median progression-free survival of 8.5 months. However, given that dovitinib does not act as a selective kinase inhibitor, it remains unclear whether the observed response in the patients was due to FGFR1 inhibition or other targeted kinases, including KIT, PDGFR, RET, CSF1-R, TrkA and FLT3. An alternative agent, lenvatinib, that inhibits not only FGFR 1–3, but also VEGFR2, KIT proto-oncogene receptor tyrosine kinase (c-KIT), ret proto-oncogene (RET) and platelet EGFRa and b, is considered a more potent therapeutic option (71). A study by Tchekmedyian et al (59) investigating lenvatinib use confirmed a partial response in 32 patients with recurrent or metastatic ACC, with 8 patients showing >20% reduction in tumor size. Both studies on lenvatinib and dovitinib required dosage modifications due to adverse effects such as hypertension, anemia or diarrhea (59,71). Lenvatinib has received a category 2B recommendation for patients with progressive, recurrent or metastatic ACC in the NCCN guidelines (8). A clinical trial combining lenvatinib and pembrolizumab did not show an improved overall response rate compared with lenvatinib alone in patients with ACC. A complete response rate was found in only 1 out of 17 patients, with 13 patients achieving stable disease (75). Another study evaluating the effectiveness of the FGFR1 inhibitor AZD4547 in treating ACC of the lacrimal gland found that adding cisplatin led to lower cell proliferation and migration compared with the control group treated with cisplatin alone (76).

c-KIT

Although receptor tyrosine kinase c-KIT is upregulated in 65–90% of ACC tumors (77), clinical trials involving imatinib, a c-KIT inhibitor, demonstrated that the drug was largely ineffective in treating salivary gland ACC (78). While imatinib has been successful in treating gastrointestinal stromal tumors and chronic myeloid leukemia (79), in one study examining its use in ACC, only 2 out of 42 patients experienced an objective tumor response (80). Similarly, dasatinib failed to show any improvement in patients with recurrent or metastatic ACC (81). Furthermore, the combination of cisplatin with imatinib did not result in any significant improvement in response rates, as only 3 patients (10%) exhibited a partial response (82). Thus, despite the high expression of c-KIT in salivary gland ACC, the protein does not appear to play a significant role in the pathogenesis of this tumor type, rendering it an unattractive target for future therapeutic interventions.

MYB

The translocation t(6;9)(q22-23;p23-24), which leads to the fusion of the MYB proto-oncogene with the NFIB transcription factor gene, is a hallmark of ACC, as it is detected in ~50% of cases (83). However, the precise frequency of MYB-NFIB fusions in ACC remains unknown, as it varies depending on the method of detection used (for example, fluorescence in situ hybridization vs. PCR/RNA-seq) and the type of material analyzed (for example, fresh-frozen vs. formalin-fixed paraffin-embedded tissue) (84). MYB protein expression by IHC was recently demonstrated in >90% of ACCs in a large multi-institutional study (85). Moreover, MYB gene upregulation itself serves a key role in the cancer pathogenesis, as it promotes tumorigenesis by enhancing cancer stemness; however, MYB was previously thought to present a difficult therapeutic target due to its nature as a transcription factor (86). Nevertheless, the current and past studies proved that MYB inhibition has had a beneficial effect on patients with ACC. The approach to MYB inhibition may be accomplished by direct MYC degradation or by the inhibition of MYB-associated proteins (87). A study by Yusenko et al (88) confirmed that use of polyether ionophore monensin A results in the inhibition of MYB and leads to its degradation in vitro. All-trans retinoic acid (ATRA) also has an inhibitory effect on the MYB gene in ACC cells (89). Another molecule inhibiting MYB-NFIB fusion is insulin-like growth factor receptor 1 (IGFR1) (90). IGFR1, similar to EGFR, stimulates ACC proliferation as the MYB-NFIB fusion in ACC is regulated by IGF1R through an autocrine loop (90). In ACC models, linstinib targeting IGFR1, crizotinib targeting ALK or geftinib targeting EGFR resulted in tumor growth reduction (91). In a clinical trial evaluating figitumab combined with dacomitinib, and another trial assessing R1507 with sorafenib, the response rate was observed in 1 patient, while stable disease was seen in 3 patients (92,93). However, due to the development of MYB-targeted inhibitors, MYB is becoming an increasingly attractive therapeutic target. In a study conducted by Yusenko et al (94), a Bcr-TMP MYB-inhibitory particle resulted in significantly decreased ACC cell proliferation.

NOTCH

The NOTCH signaling pathway serves a pivotal role in numerous cellular processes that are crucial for cell differentiation, but it has also been implicated in the pathogenesis of several types of cancer, including ACC (95). Although NOTCH1 mutations are less common compared with MYB gene alterations, they are typically found in ~15% of ACC cases (96). Furthermore, studies suggest that NOTCH mutations in patients with ACC are associated with a poorer prognosis (97). Given the role of NOTCH signaling in regulating tumor cell behavior, acting as both an oncogene and a tumor suppressor depending on the cellular and tissue context, NOTCH inhibitors are an appealing therapeutic strategy for certain subsets of ACC. Several preclinical studies have confirmed the effectiveness of NOTCH1 inhibitors in ACC patient-derived xenograft (PDX) models (98). A previous study examined AL101 (osugacestat), a potent γ-secretase inhibitor that prevents the activation of all four NOTCH receptors (99). The antitumor activity of AL101 was demonstrated in ACC cell lines, organoids and PDX models (99). Notably, AL101 showed strong antitumor effects in both in vitro and in vivo models of ACC with activating NOTCH1 mutations that constitutively upregulated NOTCH signaling pathways (99). This provides a rationale for further clinical trials evaluating AL101 in patients with NOTCH-driven relapsed or refractory ACC. In a phase II clinical trial assessing AL101, 9 out of 77 patients experienced a partial response, while 44 had stable disease (99). In a phase I study of the pan-NOTCH inhibitor CB-103 for patients with ACC and other tumors, no partial response was observed, but 23 out of 40 patients with ACC had stable disease, with a median progression-free survival time of 2.5 months and a median overall survival yime of 18.4 months (100). A study of crenigacestat another pan-NOTCH inhibitor, demonstrated that only 1 out of 22 patients with ACC had a partial response, while 15 experienced stable disease (101). Another trial testing brontictuzumab targeting Notch1 receptor, showed favorable effects, with 2 out of 12 patients achieving a partial response and 3 achieving stable disease (102).

p53

Although p53 can be considered one of the most notable genes with mutations commonly found in numerous types of cancer, its incidence in ACC is less common, accounting for 10–20% of cases (103). However, in a previous study, increased p53 expression was observed in ~90% of ACC cases, indicating that it might serve as a potential therapeutic target (104). In the ACC PDX model, inhibiting the interaction between murine double minute 2 (MDM2) and p53 using MI-733 led to apoptosis, tumor regression and the prevention of tumor recurrence (105). Following this, a clinical trial is currently underway, assessing the efficacy of blocking MDM2-p53 with alrizomadlin (APG-115), with or without platinum-based therapy, in salivary gland cancer, including ACC, with results yet to be published (106).

Phosphatidylinositol 3-kinase (PI3K)/phosphatase and tensin homolog (PTEN)/mammalian target of rapamycin (mTOR) pathway

PI3K activates AKT by phosphorylation and via certain transcriptional factors, including mTOR (107). In a study by Yu et al (108), proteins such as p-S6, p-STAT3, PAI, EGFR and hypoxia induced factor-1α were significantly elevated in ACC samples compared with those in benign salivary lesions, such as pleomorphic adenoma and normal salivary glands. Similarly, a study by Liu et al (109) demonstrated decreased expression levels of PTEN in ACC samples, particularly in the solid subtype, compared with other salivary gland malignancies. Given that PTEN functions as a tumor suppressor, its reduced expression levels in ACC tissue might present a potential therapeutic target. In a phase II clinical trial involving everolimus, an mTOR inhibitor, among patients with ACC showing disease progression, treatment with everolimus resulted in a median progression-free survival time of 11.2 months. Of the 34 participants included, 15 showed tumor shrinkage and 27 exhibited stable disease (110). Additionally, a separate phase I study investigating the combination of everolimus with lenalidomide found that this regimen was safe and well tolerated, indicating a potential combination therapy for ACC (111).

Immune checkpoint inhibitors

Immunotherapy has transformed the treatment landscape for numerous types of cancer that previously had a poor prognosis. It is now a primary treatment option for several malignancies and is considered to be promising approach in cancer therapy (112114). The expression levels of proteins such as cytotoxic T cell antigen 4 (CTLA-4), programmed death receptor 1 (PD-1) and programmed death-ligand 1 (PD-L1), which are primary targets for immunotherapeutic treatments, is significantly low in the environment of ACC (115). As ACC is considered to be a ‘cold tumor’, the lymphocytic infiltration in its microenvironment is sparse (116). Therefore, both past and ongoing research on the use of immunotherapy in ACC has yielded unsatisfactory results (15,117). The NISCAHN trial examined the efficacy of nivolumab, a PD-1 inhibitor, in 45 patients with recurrent or metastatic ACC. The results showed an overall response rate of only 0.8%, with a progression-free survival time of 6 months for 33% of patients (118). Another study of ACC patients, where nivolumab was combined with the CTLA-4 inhibitor ipilimumab, reported an even lower overall response rate compared with nivolumab alone, at ~6% (119). Additionally, a study assessing the effects of pembrolizumab acting as an PD1 inhibitor, with or without concomitant radiotherapy, did not demonstrate any significant tumor response (120). A clinical trial of pembrolizumab with or without radiotherapy showed that 65% of patients achieved disease stabilization for an average of 11 months, but no objective responses were recorded. In a study by Mosconi et al (121), no expression of PD-1 or PD-L1 was detected in ACC samples. These findings were corroborated by similar results in a study conducted by Guazzo et al (122). By contrast, the high expression level of PD-L2 in ACC tissue has garnered increasing interest in the research field (123,124). In a clinical trial of ACC patients, the PD-1 antibody BGB-A317, which acts as an inhibitor of both PD-L1 and PD-L2, was combined with the Tet-MYB vaccine (125). It appears that the PD-L1 inhibitors enhance the antitumor effect by restoring T-cell activity and improving the immune system's ability to recognize and attack tumor cells (126).

Vaccines

In the realm of immunomodulatory therapies, the TeTMYB vaccine has been developed to target MYB. This vaccine was constructed using a full-length MYB complementary DNA bound by two potent CD4+ epitopes derived from the tetanus toxin, which was subsequently cloned into the complementary DNA vaccine vector pVAX1 (127). Previously, the TeTMYB vaccine demonstrated efficacy in targeting MYB-expressing colorectal cancer in experimental animal studies (127). Subsequently, the TeTMYB vaccine underwent a phase I clinical trial not only for colorectal cancer, but also for salivary gland ACC (125).

Protein arginine methyltransferase 5 (PRMT5)

The PRMT5 inhibitor, targets PRMT5, an enzyme responsible for the methylation of arginine residues that serves a significant role in various cellular processes, including cell cycle control, DNA repair or signal transduction (128). However, its role has also been linked to the pathogenesis of several types of cancer, including ACC (129). The involvement of PRMT5 in cancer is primarily due to its inhibition of tumor suppressor gene expression, leading to a loss of control over carcinogenesis (130). In a phase I clinical trial involving 14 patients with ACC, the PRMT5 inhibitor GSK3326595 demonstrated a partial response rate of 21% (3 out of 14 patients) (130). Another study evaluating the PRMT inhibitor PRT543 in patients with recurrent or metastatic ACC reported a median progression-free survival time of 5.9 months. With 56 patients participating, the overall response rate was 2% and disease stabilization was observed in 7% of cases (131).

Tropomyosin receptor A (TRKA)

The TRK family, a group of receptor tyrosine kinases encoded by NTRK genes, plays a crucial role in the development and proper functioning of the nervous system. TRKA, along with nerve growth factor, is associated with PNI, a phenomenon highly characteristic of ACC and responsible for late recurrences or distant metastases (132). A phase-I clinical trial is assessing the effectiveness of a small molecule, VMD-928, which acts as a TRKA inhibitor, in solid tumors including ACC; however, the results of this trial are yet to be published (133).

Prostate-specific membrane antigen (PSMA)

PSMA expression is characteristic not only of prostate cancer cells, but also for other malignant diseases, including salivary ACC (134). Under normal conditions, PSMA may be present on the surface of serous and mucous acinar cells, as well as intercalated and striated duct cells (135). Moreover, PSMA appears to be more densely distributed in the major salivary glands than in the minor salivary glands, as evidenced by the increased uptake of PSMA-ligand on diagnostic images in major salivary glands (136). A study by Klein Nulent et al (135) found that PSMA expression was observed in 94% of primary ACC cases, 80% of recurrent tumors and 90% of metastatic tumors. In ACC, PSMA is predominantly localized on the surface of cancer cells, which has driven research into PSMA-based theranostics (135). A phase II study conducted by van Boxtel et al (137) using 68Ga-PSMA PET demonstrated PSMA ligand uptake in 93% of patients with ACC. These findings led to the initiation of a phase II trial focusing on PSMA radionuclide therapy (138). Given these results, PSMA expression in ACC could serve as a potential diagnostic marker and open new avenues for innovative therapeutic approaches in the future.

Serine/threonine kinase AKT

AKT serves a pivotal role in various signaling pathways and is often dysregulated in numerous types of human cancer (139). In a study involving patients with advanced stage incurable ACC, the use of MK-2206, an allosteric inhibitor of AKT, yielded no confirmed responses (140). Of the 14 included patients, 13 patients had stable disease, while 1 patient developed disease progression. The median progression-free survival time was 9.7 months, and the median overall survival time was 18.0 months (140). Consequently, AKT inhibition by MK-2206 failed to produce a significant clinical response in patients with ACC. Additionally, in lung cancer, the activation of AKT, which inhibits transcription-dependent mechanisms of ATRA, promotes invasion and cell survival, leading to resistance against retinoic acid treatment (141). This implies that AKT could be a potential therapeutic target not only in lung cancer but also in ACC.

Cancer stem cells (CSCs)

Given the high recurrence rates and chemoresistance of ACC, treatments targeting CSCs, which function as tumor-initiating cells and drive chemoresistance, may hold potential in ACC treatment (142). Although CSCs make up ~5% of all tumor cells, they are responsible for tumor heterogeneity and the capacity for self-renewal, making them a potential target for ACC management (143). Therefore, inhibitors targeting CSCs might lead to tumor regression, typically in combination with cytotoxic therapies aimed at simultaneously reducing tumor mass. Therefore, eliminating CSCs could also help reduce recurrence rates, as these cells drive ongoing tumor renewal (144). In a preclinical PDX model of ACC, inhibiting the interaction of MDM2-p53 with a small molecule inhibitor reduced the number of CSCs and increased sensitivity to cisplatin (145). Moreover, after neoadjuvant administration of MI773 and subsequent tumor resection in a preclinical study, no recurrence was observed compared with the control group that were treated with surgical tumor removal alone (107). A preclinical study on vorinostat, a histone deacetylase inhibitor, also demonstrated a reduction in CSCs in ACC (146). Additionally, the combination of vorinostat with cisplatin showed a decreased number of CSCs, indicating its potential to sensitize ACC cells to cisplatin (147). However, in a clinical trial with vorinostat and patients with recurrent/metastatic ACC, only 2 patients had a partial response, while 27 exhibited stable disease (147). A phase II clinical trial assessing the combination of chidamide and cisplatin is currently ongoing (69). In another study that focused on stemness inhibition assessing amcasertib (BBI503), which acts as multiple serine-threonine kinases inhibitor, the disease control rate was assessed at 86%, with 79% alive in the first year of survival since diagnosis (12).

Future directions

CDKs

The expression of CDK6 is significantly elevated in ACC samples, measuring 4-fold higher by mass spectrometry compared with that in squamous cell carcinoma (SCC) samples, and 3-fold higher at the mRNA level (148). Furthermore, the expression of the p16 protein, which inhibits CDK6, was observed to be notably lower in ACC samples compared with that in SCC samples (148). Since cyclins and CDKs are key regulators of the cell cycle, with CDK6 inactivating the retinoblastoma protein that acts as a G1 phase cell cycle inhibitor (149), these findings suggest that CDK6 may serve a key role in the pathogenesis of ACC and therefore, could be considered as a potential therapeutic target for future investigation.

C-X-C chemokine receptor type 4 (CXCR4)

Chemokines serve important roles in both innate and adaptive immunity (150). CXCR4 is commonly expressed on numerous types of cancer cells, with its ligand, CXCL12, contributing to cancer progression by promoting cell proliferation, migration and metastasis (151). Under normal conditions, CXCL4 is found on the surface of mesenchymal stromal cells in the lungs, liver, lymph nodes, bone marrow and peripheral nerves (152). In a study conducted by Nulent et al (152), CXCR4 expression was observed in 81% of ACC samples. Currently available literature provides data on a number of CXCR4 antagonists, including inhibitors, antibodies and microRNAs, which have been developed to target CXCR4 (153). Nevertheless, CXCR4 antagonists have been found to have limited success in clinical trials due to cellular toxicity, and poor stability and efficacy (153). Moreover, in one study, CXCL12/CXCR4 expression was found to potentially promote PNI by inducing tumor cell differentiation into Schwann-like cells via the Twist/S100A4 axis in salivary ACC (153). However, there are currently no clinical trials assessing the efficacy of these agents in patients with ACC.

Conclusions

ACC is a rare malignancy with an indolent course and elusive pathology. Currently available treatment options exhibit limited effectiveness, as late local recurrence or distant metastasis occur in >40% of patients diagnosed with ACC. Despite numerous studies conducted thus far, a definitive therapeutic target effective in ACC treatment has yet to be identified. Furthermore, even various approaches aimed at targeting both specific genes or lymphocytic infiltration in the tumor microenvironment have shown limited success. Despite these advancements, the present review confirms that progress has been made in the search for a standard therapy for ACC, though significant challenges remain. The resistance of ACC to both past and currently tested treatment options underscores the need for further research. Furthermore, the data presented in the current review suggests that multimodal therapies might be more effective in eliciting a response in ACC compared with single-agent treatments. Limited information exists on ACC pathogenesis, partly due to previous studies that repeatedly used contaminated cell lines. It seems that tumor heterogeneity and robust immune evasion mechanisms appear to contribute to the failure of novel therapies that have been effective in other types of cancer. Consequently, considerable effort must be devoted to identifying molecular causes of ACC and potential therapeutic agents.

Acknowledgements

Not applicable.

Funding

The present review was funded by the Maria Sklodowska-Curie National Research Institute of Oncology (Warsaw, Poland).

Availability of data and materials

Not applicable.

Authors' contribution

KS, AGo, AGa were responsible for conceptualization and methodology. MD, MP and JZ performed software analysis. AGo performed data visualization and investigation. KS wrote the original draft of the manuscript. AGa and KB contributed to the conception and design of the study in addition to data acquisition, analysis, and interpretation. MD was responsible for supervision and validation. MD, JZ, AGo, MMG and KBP reviewed and edited the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

ACC

adenoid cystic carcinoma

CSC

cancer stem cell

CDK

cyclin-dependent kinase

CXCR4

C-X-C chemokine receptor type 4

CTLA-4

cytotoxic T cell antigen 4

EGFR

epidermal growth factor receptor

IGFR-1

insulin-like growth factor

mTOR

mammalian target of rapamycin

PDX

patient-derived xenograft

PI3K

phosphatidylinositol 3-kinase

PSMA

prostate-specific membrane antigen

PRMT5

protein arginine methyltransferase 5

PD-1

programmed death receptor 1

PCZ

prochlorperazine

PNI

perineural invasion

SCC

squamous cell carcinoma

TRKA

tropomyosin receptor A

VEGF

vascular endothelial growth factor

References

1 

Pushpanjali M, Sujata DN, Subramanyam SB and Jyothsna M: Adenoid cystic carcinoma: An unusual presentation. J Oral Maxillofac Pathol. 18:286–290. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Gandhi AK, Roy S, Biswas A, Bhasker S, Sharma A, Thakar A and Mohanti BK: Adenoid cystic carcinoma of head and neck: A single institutional analysis of 66 patients treated with multi-modality approach. Indian J Med Paediatr Oncol. 36:166–171. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Romani C, Lorini L, Bozzola A, Bignotti E, Tomasoni M, Ardighieri L, Bugatti M, Battocchio S, Ravaggi A, Tomasini D, et al: Functional profiles of curatively treated adenoid cystic carcinoma unveil prognostic features and potentially targetable pathways. Sci Rep. 13:18092023. View Article : Google Scholar : PubMed/NCBI

4 

Zhang M, Liu Y, Yang H, Jin F and Zheng A: Breast adenoid cystic carcinoma: A report of seven cases and literature review. BMC Surg. 22:1132022. View Article : Google Scholar : PubMed/NCBI

5 

Chen Z, Jiang J, Fan Y and Lu H: Pulmonary adenoid cystic carcinoma: Molecular characteristics and literature review. Diagn Pathol. 18:652023. View Article : Google Scholar : PubMed/NCBI

6 

Alhashemi M, Oubari S, Mohamad AH, Alhashemi M, Kabel O, Alhelue A and Fattal F: Adenoid cystic carcinoma (ACC) of the Bartholin's gland misdiagnosed three times: A case report. BMC Womens Health. 23:1562023. View Article : Google Scholar : PubMed/NCBI

7 

Godge P, Sharma S and Yadav M: Adenoid cystic carcinoma of the parotid gland. Contemp Clin Dent. 3:223–226. 2012. View Article : Google Scholar : PubMed/NCBI

8 

National Comprehensive Cancer Network (NCCN), . Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Head and Neck Cancer, Version 1.2025. NCCN, Plymouth Meeting, PA. 2024.https://www.nccn.org/professionals/physician_gls/default.aspx

9 

Choi S, Sano D, Cheung M, Zhao M, Jasser SA, Ryan AJ, Mao L, Chen WT, El-Naggar AK and Myers JN: Vandetanib inhibits growth of adenoid cystic carcinoma in an orthotopic nude mouse model. Clin Cancer Res. 14:5081–5089. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Queimado L, Lopes C, Du F, Martins C, Fonseca I, Bowcock AM, Soares J and Lovett M: In vitro transformation of cell lines from human salivary gland tumors. Int J Cancer. 81:793–798. 1999. View Article : Google Scholar : PubMed/NCBI

11 

Phuchareon J, Ohta Y, Woo JM, Eisele DW and Tetsu O: Genetic profiling reveals cross-contamination and misidentification of 6 adenoid cystic carcinoma cell lines: ACC2, ACC3, ACCM, ACCNS, ACCS and CAC2. PLoS One. 4:e60402009. View Article : Google Scholar : PubMed/NCBI

12 

Miller LE, Au V, Mokhtari TE, Goss D, Faden DL and Varvares MA: A contemporary review of molecular therapeutic targets for adenoid cystic carcinoma. Cancers (Basel). 14:9922022. View Article : Google Scholar : PubMed/NCBI

13 

Michaelides I, Künzel J, Ettl T, Beckhove P, Bohr C, Brochhausen C and Mamilos A: Adenoid cystic carcinoma of the salivary glands: A pilot study of potential therapeutic targets and characterization of the immunological tumor environment and angiogenesis. Eur Arch Otorhinolaryngol. 280:2937–2944. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Linxweiler M, Wemmert S, Braun FL, Körner S, Brust LA, Knebel M, Klamminger GG, Wagner M, Morris LGT and Kühn JP: Targeted therapy in salivary gland cancer: Prevalence of a selected panel of actionable molecular alterations in a German tertiary referral center patient cohort. Mol Diagn Ther. 1:10.1007/s40291–024-00750-w. 2024.

15 

Chae YK, Chung SY, Davis AA, Carneiro BA, Chandra S, Kaplan J, Kalyan A and Giles FJ: Adenoid cystic carcinoma: Current therapy and potential therapeutic advances based on genomic profiling. Oncotarget. 6:37117–37134. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Belulescu IC, Margaritescu C, Dumitrescu CI, DĂguci L, Munteanu C and Margaritescu OC: Adenoid cystic carcinoma of salivary gland: A ten-year single institute experience. Curr Health Sci J. 46:56–65. 2020.PubMed/NCBI

17 

Fang Y, Peng Z, Wang Y, Gao K, Liu Y, Fan R, Zhang H, Xie Z and Jiang W: Current opinions on diagnosis and treatment of adenoid cystic carcinoma. Oral Oncol. 130:1059452022. View Article : Google Scholar : PubMed/NCBI

18 

Hong J, Choi E, Kim D, Seo MK, Kang H, Park B and Kim S: Immunological subtyping of salivary gland cancer identifies histological origin-specific tumor immune microenvironment. NPJ Precis Oncol. 8:152024. View Article : Google Scholar : PubMed/NCBI

19 

Atallah S, Casiraghi O, Fakhry N, Wassef M, Uro-Coste E, Espitalier F, Sudaka A, Kaminsky MC, Dakpe S, Digue L, et al: A prospective multicentre REFCOR study of 470 cases of head and neck Adenoid cystic carcinoma: Epidemiology and prognostic factors. Eur J Cancer. 130:241–249. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Jaber MA, Hassan M, Ingafou M and Elameen AM: Adenoid cystic carcinoma of the minor salivary glands: A systematic review and meta-analysis of clinical characteristics and management strategies. J Clin Med. 13:2672024. View Article : Google Scholar : PubMed/NCBI

21 

Saleh E and Ukwas A: Adenoid cystic carcinoma of salivary glands: A ten-year review and an assessment of the current management, surgery, radiotherapy, and chemotherapy. Int J Otolaryngol. 29:74014582023.PubMed/NCBI

22 

Loap P, Vischioni B, Bonora M, Ingargiola R, Ronchi S, Vitolo V, Barcellini A, Goanta L, De Marzi L, Dendale R, et al: Biological rationale and clinical evidence of carbon ion radiation therapy for adenoid cystic carcinoma: A narrative review. Front Oncol. 11:7890792021. View Article : Google Scholar : PubMed/NCBI

23 

Powell SK, Kulakova K and Kennedy S: A review of the molecular landscape of adenoid cystic carcinoma of the lacrimal gland. Int J Mol Sci. 24:137552023. View Article : Google Scholar : PubMed/NCBI

24 

Liu J, Shao C, Tan ML, Mu D, Ferris RL and Ha PK: Molecular biology of adenoid cystic carcinoma. Head Neck. 34:1665–1677. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Zupancic M, Näsman A, Friesland S and Dalianis T: Adenoid cystic carcinoma, clinical presentation, current treatment and approaches towards novel therapies. Anticancer Res. 44:1325–1334. 2024. View Article : Google Scholar : PubMed/NCBI

26 

Persson M, Andrén Y, Mark J, Horlings HM, Persson F and Stenman G: Recurrent fusion of MYB and NFIB transcription factor genes in carcinomas of the breast and head and neck. Proc Natl Acad Sci USA. 106:18740–18744. 2009. View Article : Google Scholar : PubMed/NCBI

27 

de Morais EF, de Farias Morais HG, de Almeida Freitas R and Coletta RD: Prognostic significance of histopathological parameters for salivary gland adenoid cystic carcinoma. Dent J (Basel). 11:2622023. View Article : Google Scholar : PubMed/NCBI

28 

Hoekzema CR, Massey BL, Blumin JH, Hunt BC and Bock JM: Dysphagia due to adenoid cystic carcinoma of the base of the tongue. Ann Otol Rhinol Laryngol. 121:402–406. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Marchiano E, Chin OY, Fang CH, Park RC, Baredes S and Eloy JA: Laryngeal adenoid cystic carcinoma: A systematic review. Otolaryngol Head Neck Surg. 154:433–439. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Dantas AN, Morais EF, Macedo RA, Tinôco JM and Morais ML: Clinicopathological characteristics and perineural invasion in adenoid cystic carcinoma: A systematic review. Braz J Otorhinolaryngol. 81:329–335. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Ma C, Gao T, Ju J, Zhang Y, Ni Q, Li Y, Zhao Z, Chai J, Yang X and Sun M: Sympathetic innervation contributes to perineural invasion of salivary adenoid cystic carcinoma via the β2-adrenergic receptor. Onco Targets Ther. 21:1475–1495. 2019. View Article : Google Scholar

32 

Atram MA, Shivkumar VB and Gangane NM: Intracranial remote metastasis from adenoid cystic cancer of parotid gland: Case report and review of literature. Indian J Otolaryngol Head Neck Surg. 74 (Suppl 3):S5949–S5953. 2022. View Article : Google Scholar

33 

Zheng Y, He Y, Wu F, Liu M, Wang L and Wu J: Possible local treatment for liver metastases of adenoid cystic carcinoma (ACC): Single-centre experience and literature review. Transl Cancer Res. 9:4573–4582. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Lupinetti AD, Roberts DB, Williams MD, Kupferman ME, Rosenthal DI, Demonte F, El-Naggar A, Weber RS and Hanna EY: Sinonasal adenoid cystic carcinoma: The M. D. Anderson cancer center experience. Cancer. 110:2726–2731. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Castelnuovo P and Turri-Zanoni M: Adenoid cystic carcinoma. Adv Otorhinolaryngol. 84:197–209. 2020.PubMed/NCBI

36 

Jaso J and Malhotra R: Adenoid cystic carcinoma. Arch Pathol Lab Med. 135:511–515. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Mitani Y, Li J, Rao PH, Zhao YJ, Bell D, Lippman SM, Weber RS, Caulin C and El-Naggar AK: Comprehensive analysis of the MYB-NFIB gene fusion in salivary adenoid cystic carcinoma: Incidence, variability, and clinicopathologic significance. Clin Cancer Res. 16:4722–4731. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Skálová A, Hyrcza MD and Leivo I: Update from the 5th edition of the world health organization classification of head and neck tumors: Salivary glands. Head Neck Pathol. 16:40–53. 2022. View Article : Google Scholar : PubMed/NCBI

39 

Choi SH, Yang AJ, Yoon SO, Kim HR, Hong MH, Kim SH, Choi EC, Keum KC and Lee CG: Role of postoperative radiotherapy in resected adenoid cystic carcinoma of the head and neck. Radiat Oncol. 17:1972022. View Article : Google Scholar : PubMed/NCBI

40 

Ishida E, Ogawa T, Rokugo M, Ishikawa T, Wakamori S, Ohkoshi A, Usubuchi H, Higashi K, Ishii R, Nakanome A and Katori Y: Management of adenoid cystic carcinoma of the head and neck: A single-institute study with over 25-year follow-up. Head Face Med. 16:142020. View Article : Google Scholar : PubMed/NCBI

41 

Nakamura Y, Umekawa M, Shinya Y, Hasegawa H, Shin M, Katano A, Shinozaki-Ushiku A, Kondo K and Saito N: Stereotactic radiosurgery for skull base adenoid cystic carcinoma: A report of two cases. Surg Neurol Int. 13:5092022. View Article : Google Scholar : PubMed/NCBI

42 

Ali S, Palmer FL, Katabi N, Lee N, Shah JP, Patel SG and Ganly I: Long-term local control rates of patients with adenoid cystic carcinoma of the head and neck managed by surgery and postoperative radiation. Laryngoscope. 127:2265–2269. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Iseli TA, Karnell LH, Graham SM, Funk GF, Buatti JM, Gupta AK, Robinson RA and Hoffman HT: Role of radiotherapy in adenoid cystic carcinoma of the head and neck. J Laryngol Otol. 123:1137–1144. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Geiger JL, Ismaila N, Beadle B, Caudell JJ, Chau N, Deschler D, Glastonbury C, Kaufman M, Lamarre E, Lau HY, et al: Management of Salivary Gland Malignancy: ASCO Guideline. JCO. 39:1909–1941. 2021. View Article : Google Scholar

45 

Sahara S, Herzog AE and Nör JE: Systemic therapies for salivary gland adenoid cystic carcinoma. Am J Cancer Res. 11:4092–4110. 2021.PubMed/NCBI

46 

Papaspyrou G, Hoch S, Rinaldo A, Rodrigo JP, Takes RP, van Herpen C, Werner JA and Ferlito A: Chemotherapy and targeted therapy in adenoid cystic carcinoma of the head and neck: A review. Head Neck. 33:905–911. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Ha H, Keam B, Ock CY and Heo DS: Efficacy of cyclophosphamide, doxorubicin, and cisplatin for adenoid cystic carcinoma, and their relationship with the pre-chemotherapy tumor growth rate. Chin Clin Oncol. 9:152020. View Article : Google Scholar : PubMed/NCBI

48 

Kim HR, Lee SJ, Park S, Jung HA, Lee SH, Jeong HS, Chung MK and Ahn MJ: A single-arm, prospective, phase II study of cisplatin plus weekly docetaxel as first-line therapy in patients with metastatic or recurrent salivary gland cancer. Cancer Res Treat. 54:719–727. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Dewenter I, Otto S, Kakoschke TK, Smolka W and Obermeier KT: Recent advances, systemic therapy, and molecular targets in adenoid cystic carcinoma of the head and neck. J Clin Med. 12:14632023. View Article : Google Scholar : PubMed/NCBI

50 

Xu LH, Zhao F, Yang WW, Chen CW, Du ZH, Fu M, Ge XY and Li SL: MYB promotes the growth and metastasis of salivary adenoid cystic carcinoma. Int J Oncol. 54:1579–1590. 2019.PubMed/NCBI

51 

Lin QQ, Sun JL, Wang F, Zhang HZ, Zhou G and Xi Q: Current understanding of adenoid cystic carcinoma in the gene expression and targeted therapy. Holist Integ Oncol. 2:72023. View Article : Google Scholar

52 

Pouloudi D, Sotiriadis A, Theodorakidou M, Sarantis P, Pergaris A, Karamouzis MV and Theocharis S: The impact of angiogenesis in the most common salivary gland malignant tumors. Int J Mol Sci. 21:93352020. View Article : Google Scholar : PubMed/NCBI

53 

He Q, Zhou X, Li S, Jin Y, Chen Z, Chen D, Cai Y, Liu Z, Zhao T and Wang A: MicroRNA-181a suppresses salivary adenoid cystic carcinoma metastasis by targeting MAPK-Snai2 pathway. Biochim Biophys Acta. 1830:5258–5466. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Chen C, Choudhury S, Wangsa D, Lescott CJ, Wilkins DJ, Sripadhan P, Liu X, Wangsa D, Ried T, Moskaluk C, et al: A multiplex preclinical model for adenoid cystic carcinoma of the salivary gland identifies regorafenib as a potential therapeutic drug. Sci Rep. 7:114102017. View Article : Google Scholar : PubMed/NCBI

55 

Kim Y, Lee SJ, Lee JY, Lee SH, Sun JM, Park K, An HJ, Cho JY, Kang EJ, Lee HY, et al: Clinical trial of nintedanib in patients with recurrent or metastatic salivary gland cancer of the head and neck: A multicenter phase 2 study (Korean Cancer Study Group HN14-01). Cancer. 123:1958–1964. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Chau NG, Hotte SJ, Chen EX, Chin SF, Turner S, Wang L and Siu LL: A phase II study of sunitinib in recurrent and/or metastatic adenoid cystic carcinoma (ACC) of the salivary glands: Current progress and challenges in evaluating molecularly targeted agents in ACC. Ann Oncol. 23:1562–1570. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Wagner VP, Ferrarotto R, Vargas PA, Martins MD, Bingle CD and Bingle L: Drug-based therapy for advanced adenoid cystic carcinoma: Current landscape and challenges based on an overview of registered clinical trials. Crit Rev Oncol Hematol. 181:1038862023. View Article : Google Scholar : PubMed/NCBI

58 

Ferrarotto R, Sousa LG, Feng L, Mott F, Blumenschein G, Altan M, Bell D, Bonini F, Li K, Marques-Piubelli ML, et al: Phase II clinical trial of axitinib and avelumab in patients with recurrent/metastatic adenoid cystic carcinoma. J Clin Oncol. 41:2843–2851. 2023. View Article : Google Scholar : PubMed/NCBI

59 

Tchekmedyian V, Sherman EJ, Dunn L, Tran C, Baxi S, Katabi N, Antonescu CR, Ostrovnaya I, Haque SS, Pfister DG and Ho AL: Phase II study of lenvatinib in patients with progressive, recurrent or metastatic adenoid cystic carcinoma. J Clin Oncol. 37:1529–1537. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Locati LD, Galbiati D, Calareso G, Alfieri S, Singer S, Cavalieri S, Bergamini C, Bossi P, Orlandi E, Resteghini C, et al: Patients with adenoid cystic carcinomas of the salivary glands treated with lenvatinib: Activity and quality of life. Cancer. 126:1888–1894. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Hanna GJ, Ahn MJ, Muzaffar J, Keam B, Bowles DW, Wong DJ, Ho AL, Kim SB, Worden F, Yun T, et al: A phase II trial of rivoceranib, an oral vascular endothelial growth factor receptor 2 inhibitor, for recurrent or metastatic adenoid cystic carcinoma. Clin Cancer Res. 29:4555–4563. 2023. View Article : Google Scholar : PubMed/NCBI

62 

van Boxtel W, Uijen MJM, Krens SD, Dijkema T, Willems SM, Jonker MA, Pegge SAH, van Engen-van Grunsven ACH and van Herpen CML: Excessive toxicity of cabozantinib in a phase II study in patients with recurrent and/or metastatic salivary gland cancer. Eur J Cancer. 161:128–137. 2022. View Article : Google Scholar : PubMed/NCBI

63 

Lee SK, Kwon MS, Lee YS, Choi SH, Kim SY, Cho KJ and Nam SY: Prognostic value of expression of molecular markers in adenoid cystic cancer of the salivary glands compared with lymph node metastasis: A retrospective study. World J Surg Oncol. 11:2662012. View Article : Google Scholar

64 

Kol A, van Scheltinga AT, Pool M, Gerdes C, de Vries E and de Jong S: ADCC responses and blocking of EGFR-mediated signaling and cell growth by combining the anti-EGFR antibodies imgatuzumab and cetuximab in NSCLC cells. Oncotarget. 8:45432–45446. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Agulnik M, Cohen EW, Cohen RB, Chen EX, Vokes EE, Hotte SJ, Winquist E, Laurie S, Hayes DN, Dancey JE, et al: Phase II study of lapatinib in recurrent or metastatic epidermal growth factor receptor and/or erbB2 expressing adenoid cystic carcinoma and non adenoid cystic carcinoma malignant tumors of the salivary glands. J Clin Oncol. 25:3978–3984. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Bossi P, Miceli R, Locati LD, Ferrari D, Vecchio S, Moretti G, Denaro N, Caponigro F, Airoldi M, Moro C, et al: A randomized, phase 2 study of cetuximab plus cisplatin with or without paclitaxel for the first-line treatment of patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck. Ann Oncol. 28:2820–2826. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Huang Y, Yu T, Fu X, Chen J, Liu Y, Li C, Xia Y, Zhang Z and Li L: EGFR inhibition prevents in vitro tumor growth of salivary adenoid cystic carcinoma. BMC Cell Biol. 14:132013. View Article : Google Scholar : PubMed/NCBI

68 

Dillon PM, Chakraborty S, Moskaluk CA, Joshi PJ and Thomas CY: Adenoid cystic carcinoma: A review of recent advances, molecular targets, and clinical trials. Head Neck. 38:620–627. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Chintakuntlawar AV, Okuno SH and Price KA: Systemic therapy for recurrent or metastatic salivary gland malignancies. Cancers Head Neck. 1:112016. View Article : Google Scholar : PubMed/NCBI

70 

Chew HY, De Lima PO, Cruz JL, Banushi B, Echejoh G, Hu L, Joseph SR, Lum B, Rae J, O'Donnell JS, et al: Endocytosis inhibition in humans to improve responses to ADCC-mediating antibodies. Cell. 180:895–914.e27. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Nightingale J, Lum B, Ladwa R, Simpson F and Panizza B: Adenoid cystic carcinoma: A review of clinical features, treatment targets and advances in improving the immune response to monoclonal antibody therapy. Biochim Biophys Acta Rev Cancer. 1875:1885232021. View Article : Google Scholar : PubMed/NCBI

72 

Humtsoe JO, Kim HS, Leonard B, Ling S, Keam B, Marchionni L, Afsari B, Considine M, Favorov AV, Fertig EJ, et al: Newly identified members of fgfr1 splice variants engage in cross-talk with AXL/AKT axis in salivary adenoid cystic carcinoma. Cancer Res. 81:1001–1013. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Schvartsman G, Pinto NA, Bell D and Ferrarotto R: Salivary gland tumors: Molecular characterization and therapeutic advances for metastatic disease. Head Neck. 41:239–247. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Dillon PM, Petroni GR, Horton BJ, Moskaluk CA, Fracasso PM, Douvas MG, Varhegyi N, Zaja-Milatovic S and Thomas CY: A phase II study of dovitinib in patients with recurrent or metastatic adenoid cystic carcinoma. Clin Cancer Res. 23:4138–4145. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Mohamadpour M, Sherman EJ, Kriplani A, Fetten J, Dunn L, Michel LS, Hung KW, Baxi SS, McDonald E, Conybeare R, et al: A phase II study of lenvatinib plus pembrolizumab in patients with progressive, recurrent/metastatic adenoid cystic carcinoma. J Clin Oncol. 41:60482023. View Article : Google Scholar

76 

Doddapaneni R, Tao W, Naranjo A, Nikpoor N, Tse DT and Pelaez D: Fibroblast growth factor receptor 1 (FGFR1) as a therapeutic target in adenoid cystic carcinoma of the lacrimal gland. Oncotarget. 10:480–493. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Cros J, Sbidian E, Hans S, Roussel H, Scotte F, Tartour E, Brasnu D, Laurent-Puig P, Bruneval P, Blons H and Badoual C: Expression and mutational status of treatment-relevant targets and key oncogenes in 123 malignant salivary gland tumours. Ann Oncol. 24:2624–2629. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Pfeffer MR, Talmi Y, Catane R, Symon Z, Yosepovitch A and Levitt M: A phase II study of Imatinib for advanced adenoid cystic carcinoma of head and neck salivary glands. Oral Oncol. 43:33–36. 2007. View Article : Google Scholar : PubMed/NCBI

79 

Thanopoulou E and Judson I: The safety profile of imatinib in CML and GIST: Long-term considerations. Arch Toxicol. 86:1–12. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Hotte SJ, Winquist EW, Lamont E, MacKenzie M, Vokes E, Chen EX, Brown S, Pond GR, Murgo A and Siu LL: Imatinib mesylate in patients with adenoid cystic cancers of the salivary glands expressing c-kit: A princess margaret hospital phase II consortium study. J Clin Oncol. 23:585–590. 2005. View Article : Google Scholar : PubMed/NCBI

81 

Wong SJ, Karrison T, Hayes DN, Kies MS, Cullen KJ, Tanvetyanon T, Argiris A, Takebe N, Lim D, Saba NF, et al: Phase II trial of dasatinib for recurrent or metastatic c-KIT expressing adenoid cystic carcinoma and for nonadenoid cystic malignant salivary tumors. Ann Oncol. 27:318–323. 2016. View Article : Google Scholar : PubMed/NCBI

82 

Ghosal N, Mais K, Shenjere P, Julyan P, Hastings D, Ward T, Ryder WD, Bruce I, Homer J and Slevin NJ: Phase II study of cisplatin and imatinib in advanced salivary adenoid cystic carcinoma. Br J Oral Maxillofac Surg. 49:510–515. 2011. View Article : Google Scholar : PubMed/NCBI

83 

McIntyre JB, Ko JJ, Siever J, Chan AMY, Simpson RHW, Hao D and Lau HY: MYB-NFIB gene fusions identified in archival adenoid cystic carcinoma tissue employing NanoString analysis: An exploratory study. Diagn Pathol. 14:782019. View Article : Google Scholar : PubMed/NCBI

84 

de Almeida-Pinto YD, Costa SFDS, de Andrade BAB, Altemani A, Vargas PA, Abreu LG and Fonseca FP: t(6;9)(MYB-NFIB) in head and neck adenoid cystic carcinoma: A systematic review with meta-analysis. Oral Dis. 25:1277–1282. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Persson M, Andersson MK, Mitani Y, Brandwein-Weber MS, Frierson HF Jr, Moskaluk C, Fonseca I, Ferrarotto R, Boecker W, Loening T, et al: Rearrangements, expression, and clinical significance of MYB and MYBL1 in adenoid cystic carcinoma: A multi-institutional study. Cancers (Basel). 14:36912022. View Article : Google Scholar : PubMed/NCBI

86 

Cicirò Y and Sala A: MYB oncoproteins: Emerging players and potential therapeutic targets in human cancer. Oncogenesis. 10:192021. View Article : Google Scholar : PubMed/NCBI

87 

Clesham K, Walf-Vorderwülbecke V, Gasparoli L, Virely C, Cantilena S, Tsakaneli A, Inglott S, Adams S, Samarasinghe S, Bartram J, et al: Identification of a c-MYB-directed therapeutic for acute myeloid leukemia. Leukemia. 36:1541–1549. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Yusenko MV, Trentmann A, Andersson MK, Ghani LA, Jakobs A, Paz MF, Mikesch JH, von Kries JP, Stenman G and Klempnauer KH: Monensin, a novel potent MYB inhibitor, suppresses proliferation of acute myeloid leukemia and adenoid cystic carcinoma cells. Cancer Lett. 479:61–70. 2020. View Article : Google Scholar : PubMed/NCBI

89 

Klempnauer KH: Transcription factor MYB as therapeutic target: Current developments. Int J Mol Sci. 25:32312024. View Article : Google Scholar : PubMed/NCBI

90 

Andersson MK, Åman P and Stenman G: IGF2/IGF1R signaling as a therapeutic target in MYB-positive adenoid cystic carcinomas and other fusion gene-driven tumors. Cells. 8:9132019. View Article : Google Scholar : PubMed/NCBI

91 

Andersson MK, Afshari MK, Andrén Y, Wick MJ and Stenman G: Targeting the oncogenic transcriptional regulator MYB in adenoid cystic carcinoma by inhibition of IGF1R/AKT signaling. J Natl Cancer Inst. 1:1092017.

92 

Calvo E, Soria JC, Ma WW, Wang T, Bahleda R, Tolcher AW, Gernhardt D, O'Connell J, Millham R, Giri N, et al: A phase I clinical trial and independent patient-derived xenograft study of combined targeted treatment with dacomitinib and figitumumab in advanced solid tumors. Clin Cancer Res. 23:1177–1185. 2017. View Article : Google Scholar : PubMed/NCBI

93 

Mahadevan D, Sutton GR, Arteta-Bulos R, Bowden CJ, Miller PJ, Swart RE, Walker MS, Haluska P, Munster PN, Marshall J, et al: Phase 1b study of safety, tolerability and efficacy of R1507, a monoclonal antibody to IGF-1R in combination with multiple standard oncology regimens in patients with advanced solid malignancies. Cancer Chemother Pharmacol. 73:467–473. 2014. View Article : Google Scholar : PubMed/NCBI

94 

Yusenko MV, Biyanee A, Frank D, Köhler LHF, Andersson MK, Khandanpour C, Schobert R, Stenman G, Biersack B and Klempnauer KH: Bcr-TMP, a novel nanomolar-active compound that exhibits both MYB- and microtubule-inhibitory activity. Cancers (Basel). 14:432021. View Article : Google Scholar : PubMed/NCBI

95 

Feeney L, Hapuarachi B, Adderley H, Rack S, Morgan D, Walker R, Rauch R, Herz E, Kaye J, Harrington K and Metcalf R: Clinical disease course and survival outcomes following disease recurrence in adenoid cystic carcinoma with and without NOTCH signaling pathway activation. Oral Oncol. 133:1060282022. View Article : Google Scholar : PubMed/NCBI

96 

Ferrarotto R and Heymach JV: Taking it up a NOTCH: A novel subgroup of ACC is identified. Oncotarget. 8:81725–81726. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Ferrarotto R, Mitani Y, Diao L, Guijarro I, Wang J, Zweidler-McKay P, Bell D, William WN Jr, Glisson BS, Wick MJ, et al: Activating NOTCH1 mutations define a distinct subgroup of patients with adenoid cystic carcinoma who have poor prognosis, propensity to bone and liver metastasis, and potential responsiveness to notch1 inhibitors. J Clin Oncol. 35:352–360. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Sun B, Wang Y, Sun J, Zhang C, Xia R, Xu S, Sun S and Li J: Establishment of patient-derived xenograft models of adenoid cystic carcinoma to assess pre-clinical efficacy of combination therapy of a PI3K inhibitor and retinoic acid. Am J Cancer Res. 11:773–792. 2021.PubMed/NCBI

99 

Ferrarotto R, Wirth LJ, Muzaffar J, Rodriguez CP, Xia B, Perez CA, Bowles DW, Winquist E, Hotte SJ, Metcalf R, et al: 919MO ACCURACY a phase II trial of AL101, a selective gamma secretase inhibitor, in subjects with recurrent/metastatic (R/M) adenoid cystic carcinoma (ACC) harboring Notch activating mutations (Notchmut). Ann Oncol. 31:S6632020. View Article : Google Scholar

100 

Hanna GJ, Stathis A, Lopez-Miranda E, Racca F, Quon D, Leyvraz S, Hess D, Keam B, Rodon J, Ahn MJ, et al: A phase I study of the pan-notch inhibitor CB-103 for patients with advanced adenoid cystic carcinoma and other tumors. Cancer Res Commun. 3:1853–1861. 2023. View Article : Google Scholar : PubMed/NCBI

101 

Doi T, Tajimi M, Mori J, Asou H, Inoue K, Benhadji KA and Naito Y: A phase 1 study of crenigacestat (LY3039478), the Notch inhibitor, in Japanese patients with advanced solid tumors. Invest New Drugs. 39:469–476. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Ferrarotto R, Eckhardt G, Patnaik A, LoRusso P, Faoro L, Heymach JV, Kapoun AM, Xu L and Munster P: A phase I dose-escalation and dose-expansion study of brontictuzumab in subjects with selected solid tumors. Ann Oncol. 29:1561–1568. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Li Q, Huang P, Zheng C, Wang J and Ge M: Prognostic significance of p53 immunohistochemical expression in adenoid cystic carcinoma of the salivary glands: A meta-analysis. Oncotarget. 8:29458–29473. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Carlinfante G, Lazzaretti M, Ferrari S, Bianchi B and Crafa P: P53, bcl-2 and Ki-67 expression in adenoid cystic carcinoma of the palate. A clinico-pathologic study of 21 cases with long-term follow-up. Pathol Res Pract. 200:791–799. 2005. View Article : Google Scholar : PubMed/NCBI

105 

Nör F, Warner KA, Zhang Z, Acasigua GA, Pearson AT, Kerk SA, Helman JI, Filho M, Wang S and Nör JE: Therapeutic inhibition of the MDM2-p53 interaction prevents recurrence of adenoid cystic carcinomas. Clin Cancer Res. 23:1036–1048. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Swiecicki P, Tang Y, Pearson AT, Muzaffar J, Fidler MJ, Winkler R, Fu T, Li M, Ahmad M, Yang D and Zhai Y: Trial in progress: A phase I/II trial of novel MDM2 inhibitor alrizomadlin (APG-115), with or without platinum chemotherapy, in patients with p53 wild-type salivary gland carcinoma. J Clin Oncol. 39:TPS6094. 2021. View Article : Google Scholar

107 

He Y, Sun MM, Zhang GG, Yang J, Chen KS, Xu WW and Li B: Targeting PI3K/Akt signal transduction for cancer therapy. Sig Transduct Target Ther. 6:4252021. View Article : Google Scholar : PubMed/NCBI

108 

Yu GT, Bu LL, Zhao YY, Liu B, Zhang WF, Zhao YF, Zhang L and Sun ZJ: Inhibition of mTOR reduce Stat3 and PAI related angiogenesis in salivary gland adenoid cystic carcinoma. Am J Cancer Res. 4:764–775. 2014.PubMed/NCBI

109 

Liu H, Du L, Wang R, Wei C, Liu B, Zhu L, Liu P, Liu Q, Li J, Lu SL and Xiao J: High frequency of loss of PTEN expression in human solid salivary adenoid cystic carcinoma and its implication for targeted therapy. Oncotarget. 6:11477–11491. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Kim DW, Oh DY, Shin SH, Kang JH, Cho BC, Chung JS, Kim H, Park KU, Kwon JH, Han JY, et al: A multicenter phase II study of everolimus in patients with progressive unresectable adenoid cystic carcinoma. BMC Cancer. 14:7952014. View Article : Google Scholar : PubMed/NCBI

111 

Harvey RD, Carthon BC, Lewis C, Hossain MS, Zhang C, Chen Z, Harris WB, Alese OB, Shaib W, Bilen MA, et al: Phase 1 safety and pharmacodynamic study of lenalidomide combined with everolimus in patients with advanced solid malignancies with efficacy signal in adenoid cystic carcinoma. Br J Cancer. 123:1228–1234. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Garg P, Pareek S, Kulkarni P, Horne D, Salgia R and Singhal SS: Next-Generation immunotherapy: Advancing clinical applications in cancer treatment. J Clin Med. 13:65372024. View Article : Google Scholar : PubMed/NCBI

113 

Liu B, Zhou H, Tan L, Siu KTH and Guan XY: Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther. 9:1752024. View Article : Google Scholar : PubMed/NCBI

114 

Waldman AD, Fritz JM and Lenardo MJ: A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat Rev Immunol. 20:651–668. 2020. View Article : Google Scholar : PubMed/NCBI

115 

Wolkow N, Jakobiec FA, Afrogheh AH, Kidd M, Eagle RC, Pai SI and Faquin WC: PD-L1 and PD-L2 expression levels are low in primary and secondary adenoid cystic carcinomas of the orbit: Therapeutic implications. Ophthalmic Plast Reconstr Surg. 36:444–450. 2020. View Article : Google Scholar : PubMed/NCBI

116 

Dou S, Li R, He N, Zhang M, Jiang W, Ye L, Yang Y, Zhao G, Yang Y, Li J, et al: The immune landscape of chinese head and neck adenoid cystic carcinoma and clinical implication. Front Immunol. 12:6183672021. View Article : Google Scholar : PubMed/NCBI

117 

Sato R, Yamaki H, Komatsuda H, Wakisaka R, Inoue T, Kumai T and Takahara M: Exploring immunological effects and novel immune adjuvants in immunotherapy for salivary gland cancers. Cancers (Basel). 16:12052024. View Article : Google Scholar : PubMed/NCBI

118 

Fayette J, Even C, Digue L, Geoffrois L, Rolland F, Cupissol D, Guigay J, Tourneau CL, Dillies AF, Zanetta S, et al: NISCAHN: A phase II, multicenter nonrandomized trial aiming at evaluating nivolumab (N) in two cohorts of patients (pts) with recurrent/metastatic (R/M) salivary gland carcinoma of the head and neck (SGCHN), on behalf of the unicancer head & neck group. J Clin Oncol. 37:60832019. View Article : Google Scholar

119 

Vos JL, Burman B, Jain S, Fitzgerald CWR, Sherman EJ, Dunn LA, Fetten JV, Michel LS, Kriplani A, Ng KK, et al: Nivolumab plus ipilimumab in advanced salivary gland cancer: A phase 2 trial. Nat Med. 29:3077–3089. 2023. View Article : Google Scholar : PubMed/NCBI

120 

Mahmood U, Bang A, Chen YH, Mak RH, Lorch JH, Hanna GJ, Nishino M, Manuszak C, Thrash EM, Severgnini M, et al: A randomized phase 2 study of pembrolizumab with or without radiation in patients with recurrent or metastatic adenoid cystic carcinoma. Int J Radiat Oncol Biol Phys. 109:134–144. 2021. View Article : Google Scholar : PubMed/NCBI

121 

Mosconi C, de Arruda JAA, de Farias ACR, Oliveira GAQ, de Paula HM, Fonseca FP, Mesquita RA, Silva TA, Mendonça EF and Batista AC: Immune microenvironment and evasion mechanisms in adenoid cystic carcinomas of salivary glands. Oral Oncol. 88:95–101. 2019. View Article : Google Scholar : PubMed/NCBI

122 

Guazzo E, Cooper C, Wilkinson L, Feng S, King B, Simpson F, Porceddu S, Panizza B and Coward JIG: Therapeutic implications of immune-profiling and EGFR expression in salivary gland carcinoma. Head Neck. 43:768–777. 2021. View Article : Google Scholar : PubMed/NCBI

123 

Chang H, Kim JS, Choi YJ, Cho JG, Woo JS, Kim A, Kim JS and Kang EJ: Overexpression of PD-L2 is associated with shorter relapse-free survival in patients with malignant salivary gland tumors. Onco Targets Ther. 10:2983–2992. 2017. View Article : Google Scholar : PubMed/NCBI

124 

Muraro E, Romanò R, Fanetti G, Vaccher E, Turturici I, Lupato V, La Torre FB, Polesel J, Fratta E, Giacomarra V, et al: Tissue and circulating PD-L2: Moving from health and immune-mediated diseases to head and neck oncology. Crit Rev Oncol Hematol. 175:1037072022. View Article : Google Scholar : PubMed/NCBI

125 

Pham T, Pereira L, Roth S, Galletta L, Link E, Akhurst T, Solomon B, Michael M, Darcy P, Sampurno S, et al: First-in-human phase I clinical trial of a combined immune modulatory approach using TetMYB vaccine and Anti-PD-1 antibody in patients with advanced solid cancer including colorectal or adenoid cystic carcinoma: The MYPHISMO study protocol (NCT03287427). Contemp Clin Trials Commun. 16:1004092019. View Article : Google Scholar : PubMed/NCBI

126 

Parvez A, Choudhary F, Mudgal P, Khan R, Qureshi KA, Farooqi H and Aspatwar A: PD-1 and PD-L1: Architects of immune symphony and immunotherapy breakthroughs in cancer treatment. Front Immunol. 14:12963412023. View Article : Google Scholar : PubMed/NCBI

127 

Cross RS, Malaterre J, Davenport AJ, Carpinteri S, Anderson RL, Darcy PK and Ramsay RG: Therapeutic DNA vaccination against colorectal cancer by targeting the MYB oncoprotein. Clin Transl Immunology. 4:e302015. View Article : Google Scholar : PubMed/NCBI

128 

Shailesh H, Zakaria ZZ, Baiocchi R and Sif S: Protein arginine methyltransferase 5 (PRMT5) dysregulation in cancer. Oncotarget. 9:36705–36718. 2018. View Article : Google Scholar : PubMed/NCBI

129 

Feustel K and Falchook GS: Protein arginine methyltransferase 5 (PRMT5) inhibitors in oncology clinical trials: A review. J Immunother Precis Oncol. 5:58–67. 2022. View Article : Google Scholar : PubMed/NCBI

130 

Siu L, Rasco D, Vinay SP, Romano PM, Menis J, Opdam F, Heinhuis K, Egger J, Gorman S, Parasrampuria R, et al: METEOR-1: A phase I study of GSK3326595, a first-in-class protein arginine methyltransferase 5 (PRMT5) inhibitor, in advanced solid tumours. Ann Oncol. 30:v1592019. View Article : Google Scholar

131 

Ferrarotto R, Swiecicki PL, Zandberg DP, Baiocchi RA, Wesolowski R, Rodriguez CP, McKean M, Kang H, Monga V, Nath R, et al: PRT543, a protein arginine methyltransferase 5 inhibitor, in patients with advanced adenoid cystic carcinoma: An open-label, phase I dose-expansion study. Oral Oncol. 149:1066342024. View Article : Google Scholar : PubMed/NCBI

132 

Long Y, Yao DS, Wei YS and Wu GT: Effects of nerve growth factor expression on perineural invasion and worse prognosis in early-stage cervical cancer. Chin Med J (Engl). 131:2360–2363. 2018. View Article : Google Scholar : PubMed/NCBI

133 

Chung V, Wang L, Fletcher MS, Massarelli E, Cristea MC, Kamaraju S, Alistar AT, Feng C, Li Y, Whiting RL, et al: First-time in-human study of VMD-928, an oral allosteric TrkA selective inhibitor targeting TrkA protein overexpression, in patients with solid tumors or lymphoma. J Clin Oncol. 39:30812021. View Article : Google Scholar

134 

Sivrice ME, Yasan H, Kumbul YÇ, Ertunç O and Sayın S: The importance of prostate-specific membrane antigen expression in salivary gland tumors. Turk Arch Otorhinolaryngol. 60:206–211. 2022. View Article : Google Scholar : PubMed/NCBI

135 

Nulent TJW, Valstar MH, Smit LA, Smeele LE, Zuithoff NPA, de Keizer B, de Bree R, van Es RJJ and Willems SM: Prostate-specific membrane antigen (PSMA) expression in adenoid cystic carcinoma of the head and neck. BMC Cancer. 20:5192020. View Article : Google Scholar : PubMed/NCBI

136 

Lauri C, Chiurchioni L, Russo VM, Zannini L and Signore A: PSMA expression in solid tumors beyond the prostate gland: Ready for theranostic applications? J Clin Med. 11:65902022. View Article : Google Scholar : PubMed/NCBI

137 

van Boxtel W, Lütje S, van Engen-van Grunsven ICH, Verhaegh GW, Schalken JA, Jonker MA, Nagarajah J, Gotthardt M and van Herpen CML: 68Ga-PSMA-HBED-CC PET/CT imaging for adenoid cystic carcinoma and salivary duct carcinoma: A phase 2 imaging study. Theranostics. 10:2273–2283. 2020. View Article : Google Scholar : PubMed/NCBI

138 

Tan BF, Tan WCC, Wang FQ, Lechner M, Schartinger VH, Tan DSW, Loke KSH and Nei WL: PSMA PET imaging and therapy in adenoid cystic carcinoma and other salivary gland cancers: A systematic review. Cancers (Basel). 14:35852022. View Article : Google Scholar : PubMed/NCBI

139 

Revathidevi S and Munirajan AK: Akt in cancer: Mediator and more. Semin Cancer Biol. 59:80–91. 2019. View Article : Google Scholar : PubMed/NCBI

140 

Ho AL, Foster NR, Vasudeva SD, Katabi N, Antonescu CR, Frenette GP, Pfister DG, Erlichman C and Schwartz GK: A phase 2 study of MK-2206 in patients with incurable adenoid cystic carcinoma. Cancer. 130:702–712. 2024. View Article : Google Scholar : PubMed/NCBI

141 

García-Regalado A, Vargas M, García-Carrancá A, Aréchaga-Ocampo E and González-De la Rosa CH: Activation of Akt pathway by transcription-independent mechanisms of retinoic acid promotes survival and invasion in lung cancer cells. Mol Cancer. 12:442013. View Article : Google Scholar : PubMed/NCBI

142 

Sun HR, Wang S, Yan SC, Zhang Y, Nelson PJ, Jia HL, Qin LX and Dong QZ: Therapeutic strategies targeting cancer stem cells and their microenvironment. Front Oncol. 9:11042019. View Article : Google Scholar : PubMed/NCBI

143 

Adams A, Warner K and Nör JE: Salivary gland cancer stem cells. Oral Oncol. 49:845–853. 2013. View Article : Google Scholar : PubMed/NCBI

144 

Cojoc M, Mäbert K, Muders MH and Dubrovska A: A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin Cancer Biol. 31:16–27. 2015. View Article : Google Scholar : PubMed/NCBI

145 

Sun S and Wang Z: ALDH high adenoid cystic carcinoma cells display cancer stem cell properties and are responsible for mediating metastasis. Biochem Biophys Res Commun. 396:843–848. 2010. View Article : Google Scholar : PubMed/NCBI

146 

Almeida LO, Guimarães DM, Martins MD, Martins MAT, Warner KA, Nör JE, Castilho RM and Squarize CH: Unlocking the chromatin of adenoid cystic carcinomas using HDAC inhibitors sensitize cancer stem cells to cisplatin and induces tumor senescence. Stem Cell Res. 21:94–105. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Manou M, Kanakoglou DS, Loupis T, Vrachnos DM, Theocharis S, Papavassiliou AG and Piperi C: Role of histone deacetylases in the pathogenesis of salivary gland tumors and therapeutic targeting options. Int J Mol Sci. 24:100382023. View Article : Google Scholar : PubMed/NCBI

148 

Topacio BR, Zatulovskiy E, Cristea S, Xie S, Tambo CS, Rubin SM, Sage J, Kõivomägi M and Skotheim JM: Cyclin D-Cdk4,6 drives cell-cycle progression via the retinoblastoma protein's C-Terminal Helix. Mol Cell. 74:758–770. 2019. View Article : Google Scholar : PubMed/NCBI

149 

Esche C, Stellato C and Beck LA: Chemokines: Key players in innate and adaptive immunity. J Invest Dermatol. 125:615–628. 2005. View Article : Google Scholar : PubMed/NCBI

150 

Shi Y, Riese DJ II and Shen J: The role of the CXCL12/CXCR4/CXCR7 chemokine axis in cancer. Front Pharmacol. 11:5746672020. View Article : Google Scholar : PubMed/NCBI

151 

Sun X, Cheng G, Hao M, Zheng J, Zhou X, Zhang J, Taichman RS, Pienta KJ and Wang J: CXCL12/CXCR4/CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev. 29:709–722. 2010. View Article : Google Scholar : PubMed/NCBI

152 

Nulent TJW, van Es RJJ, Valstar MH, Smeele LE, Smit LA, Gunnewiek RK, Zuithoff NPA, de Keizer B, de Bree R and Willems SM: High CXCR4 expression in adenoid cystic carcinoma of the head and neck is associated with increased risk of locoregional recurrence. J Clin Pathol. 73:476–482. 2020. View Article : Google Scholar : PubMed/NCBI

153 

Hamshaw I, Cominetti MD, Lai WY, Searcey M and Mueller A: The development of potent, competitive CXCR4 antagonists for the prevention of cancer metastasis. Biochem Pharmacol. 218:1159212023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2025
Volume 29 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Stawarz K, Durzynska M, Gałązka A, Gorzelnik A, Zwolinski J, Paszkowska M, Bieńkowska‑Pluta K and Misiak‑Galazka M: Current landscape and future directions of therapeutic approaches for adenoid cystic carcinoma of the salivary glands (Review). Oncol Lett 29: 153, 2025.
APA
Stawarz, K., Durzynska, M., Gałązka, A., Gorzelnik, A., Zwolinski, J., Paszkowska, M. ... Misiak‑Galazka, M. (2025). Current landscape and future directions of therapeutic approaches for adenoid cystic carcinoma of the salivary glands (Review). Oncology Letters, 29, 153. https://doi.org/10.3892/ol.2025.14899
MLA
Stawarz, K., Durzynska, M., Gałązka, A., Gorzelnik, A., Zwolinski, J., Paszkowska, M., Bieńkowska‑Pluta, K., Misiak‑Galazka, M."Current landscape and future directions of therapeutic approaches for adenoid cystic carcinoma of the salivary glands (Review)". Oncology Letters 29.3 (2025): 153.
Chicago
Stawarz, K., Durzynska, M., Gałązka, A., Gorzelnik, A., Zwolinski, J., Paszkowska, M., Bieńkowska‑Pluta, K., Misiak‑Galazka, M."Current landscape and future directions of therapeutic approaches for adenoid cystic carcinoma of the salivary glands (Review)". Oncology Letters 29, no. 3 (2025): 153. https://doi.org/10.3892/ol.2025.14899